throbber
J. vet. Pharmacol. Therap. 35 (Suppl. 2), 45–51. doi: 10.1111/j.1365-2885.2012.01409.x
`
`Naloxone reversal of an overdose of a novel, long-acting transdermal
`fentanyl solution in laboratory Beagles
`
`K. J. FREISE*
`
`G. C. NEWBOUND*

`
`C. TUDAN
`
`&
`
`T. P. CLARK*
`
`*Nexcyon Pharmaceuticals, Inc., Madison,

`WI, USA;
`BioAccurate Enterprises, Inc.
`Vancouver, BC, Canada
`
`Freise, K. J., Newbound, G. C., Tudan, C., Clark, T. P. Naloxone reversal of an
`overdose of a novel, long-acting transdermal fentanyl solution in laboratory
`Beagles. J. vet. Pharmacol. Therap. 35 (Suppl. 2), 45–51.
`
`Opioid overdose in dogs is manifested by clinical signs such as excessive
`sedation, bradycardia, and hypothermia. The ability of two different intramus-
`cular (i.m.) naloxone reversal regimens to reverse the opioid-induced effects of a
`fivefold overdose of long-acting transdermal fentanyl solution was evaluated in
`dogs. Twenty-four healthy Beagles were administered a single 13 mg ⁄ kg dose
`(fivefold overdose) of transdermal fentanyl solution and randomized to two
`naloxone reversal regimen treatment groups, hourly administration for 8 h of
`40 (n = 8) or 160 lg ⁄ kg i.m. (n = 16). All dogs were sedated and had reduced
`body temperatures and heart rates (HRs) prior to naloxone administration.
`Both dosage regimens significantly reduced sedation (P < 0.001), and the
`160 lg ⁄ kg naloxone regimen resulted in a nearly threefold lower odds of
`the 40 lg ⁄ kg i.m. naloxone regimen (P < 0.05).
`sedation than that of
`Additionally, naloxone significantly increased the mean body temperatures
`and HR (P < 0.001), although the 160 lg ⁄ kg regimen increased body
`temperature and HR more (P < 0.05). However, the narcotic side effects of
`fentanyl returned within 1–3 h following termination of the naloxone dosage
`regimens. The opioid-induced effects of an overdose of transdermal fentanyl
`solution can be safely and effectively reversed by either 40 or 160 lg ⁄ kg i.m.
`naloxone administered at hourly intervals.
`
`(Paper received 30 January 2012; accepted for publication 16 April 2012)
`
`Dr Terrence Clark, Nexcyon Pharmaceuticals, Inc., 644 W. Washington Ave.,
`Madison, WI 53703, USA. E-mail: clarktp@nexcyon. com
`
`INTRODUCTION
`
`The general recommendation for the selection and use of
`postoperative analgesics depends on the anticipated magnitude
`and duration of pain which in turn is influenced by the site,
`nature, and extent of surgery. In addition, both the character-
`istics of the analgesic and patient factors must be considered in
`the selection and continued use of an analgesic. The character-
`istics of an ideal analgesic have been considered and may
`include,
`in part, that the agent is a full agonist providing
`maximal analgesia for a wide range of pain states; has a rapid
`onset of action and a long duration of action; has linear kinetics;
`produces minimal adverse effects; is not vulnerable to important
`drug–drug interactions;
`is not significantly bound to plasma
`proteins; has no active metabolites; and it is reversible (Smith,
`2008; Moore, 2009). Many strategies have been pursued to
`identify technologies that meet these characteristics. At the drug
`discovery level, attempts have been made to identify an ideal
`analgesic by engineering the mu-opioid receptor (Tao et al.,
`
`Ó 2012 Blackwell Publishing Ltd
`
`2010). Others have concentrated on ways to alter existing
`analgesics or to combine existing analgesic compounds with
`drugs that may improve effectiveness while minimizing adverse
`effects (Smith, 2008).
`ideal analgesics and are
`Opioids have some features of
`generally regarded as an important part of multimodal
`postoperative analgesia, especially for moderate-to-severe pain.
`In veterinary medicine, there are limited approved products and
`extended extra-label use of orally administered opioids in dogs
`beyond the immediate postoperative period is prevented by
`inherent
`limitations including poor oral bioavailability and
`rapid clearance (Pascoe, 2000). As a result, extra-label opioid
`use is primarily limited to preoperative epidural or intrathecal
`injections, single or repeat parenteral
`injections or constant
`rate intravenous infusions delivered during anesthesia. To
`overcome these limitations and prolong the therapeutic dura-
`tion of action, other variations
`in opioid pharmaceutical
`delivery have been advanced for human or veterinary use that
`include extended release oral
`tablets
`(Holt
`et al., 2007),
`
`45
`
`Nalox1030
`Nalox-1 Pharmaceuticals, LLC
`Page 1 of 7
`
`

`

`46 K. J. Freise et al.
`
`transdermal patches (Hofmeister & Egger, 2004), and liposome-
`encapsulated injectable opioids (Smith et al., 2004). As a
`delivery method, the transdermal route has several potential
`strengths over oral and parenteral dose. These include nonin-
`vasive dosing, avoidance of the gastrointestinal tract, lack of
`first pass metabolism, steady, continuous drug delivery rather
`than a peak and trough phenomenon, potential reduction of
`side effects by elimination of peaks, possible reduction of lack of
`effectiveness owing to the elimination of troughs, and reduced
`dose frequency for convenience and increased compliance
`(Urquhart, 2000).
`Recently, a novel, long-acting transdermal fentanyl solution
`(RecuvyraÔ 50 mg ⁄ mL transdermal solution; Nexcyon Phar-
`maceuticals Ltd, London, UK) has been developed that
`potentially mitigates the disadvantages of oral, parenteral,
`and patch-delivered opioids and has several features of an ideal
`analgesic that include the following:
`fentanyl
`is a selective,
`l-opioid receptor agonist with a potency 100 times that of
`morphine (Stanley, 1992); it has a rapid onset of action and
`long duration of action, providing analgesic concentrations of
`fentanyl within 2–4 h of application for a duration of at
`least 4 days
`(Freise et al., 2012a,b);
`it has demonstrated
`dose-proportional plasma fentanyl concentrations following a
`single topical application (Freise et al., 2012a);
`there are
`minimum adverse effects at the selected dose with well-known
`opioid adverse events increasing in magnitude and frequency
`when administered up to five times the dose (Savides et al.,
`2012).
`As an ideal drug feature, reversibility has not been examined
`with transdermal
`fentanyl solution. Reversibility allows clini-
`cians to terminate the clinical effects of a drug when they are no
`longer deemed necessary to case management and permits
`intervention in the event of an overdose. Naloxone is an FDA
`approved opioid antagonist (NADA 035-825) that is considered
`the fentanyl reversal agent of choice in dogs because, as a pure
`opioid receptor competitive antagonist,
`it does not have the
`respiratory side effects of other opioid antagonists (Adams, 2001;
`Plumb, 2002). It has the highest affinity at the l-opioid receptor
`and successfully reverses the effects of fentanyl citrate injections
`in the dog (Paddleford & Short, 1973; Veng-Pedersen et al.,
`1995; Adams, 2001).
`The outcome of a fivefold overdose of transdermal fentanyl
`solution has been described and includes, in part, moderate-to-
`severe sedation, reduced rectal temperature, and reduced HR
`(Savides et al., 2012). It is likely that opioid-induced adverse
`events are reversible; however, given the long duration of action
`of transdermal fentanyl solution, it remains to be determined the
`duration of reversal
`from a single injection of naloxone.
`Therefore, the objective of this study was to determine an
`intramuscular (i.m.) naloxone reversal regimen to the opioid-
`induced effects from an overdose of
`transdermal
`fentanyl
`solution in dogs. To achieve the objective, two different i.m.
`naloxone doses were administered at hourly intervals and were
`evaluated for the reversal of peak sedation, reduced rectal
`temperature, and reduced HR in Beagle dogs following the
`
`administration of a single fivefold overdose (13 mg ⁄ kg) of
`transdermal fentanyl solution.
`
`MATERIALS AND METHODS
`
`Animals and experimental design
`
`Twenty-four healthy (based on physical examination) purpose-
`bred laboratory Beagles (12 males ⁄ 12 females), 5–6 months of
`age and ranging in bodyweight from 4.35 to 8.20 kg were
`selected. Dogs were individually housed, fed a commercial dry
`food formula and allowed ad libitum access to water. The animal
`facility temperature was maintained between 18 and 29 °C with
`30–70% relative humidity. The 24 selected dogs were random-
`ized to two different i.m. naloxone treatment groups, 40 lg ⁄ kg
`(n = 8) and 160 lg ⁄ kg (n = 16). An unbalanced study design
`was utilized because based on pilot experiments it was suspected
`that the recommended 40 lg ⁄ kg i.m. naloxone dose (Plumb,
`2002) would not provide sufficient reversal at the administered
`dose of transdermal fentanyl solution. All dogs were administered
`a single fivefold (13.0 mg ⁄ kg) overdose (use dose of 2.6 mg ⁄ kg)
`of transdermal fentanyl solution (RecuvyraÔ 50 mg ⁄ mL trans-
`dermal solution; Nexcyon Pharmaceuticals Ltd) to the ventral
`abdomen using a proprietary applicator tip as previously
`described (Freise et al., 2012a). Prior studies demonstrated that
`plasma fentanyl concentrations were as high or higher with
`ventral abdomen application compared to the labeled dorsal,
`interscalpular region application (Freise et al., 2012a). As this
`was an intentional overdose study, the worse case was examined
`with the ventral abdomen application. Sixteen hours after the
`application of transdermal fentanyl solution, when near maximal
`side effects were expected to be obtained (Savides et al., 2012),
`i.m. naloxone was administered into the dorsal lumbar muscles
`hourly for eight doses according to the treatment randomization.
`Sedation assessments were conducted by blinded assessors as
`none or sedated. There was no attempt to distinguish mild,
`moderate, or severe sedation because a previous study with a
`fivefold transdermal
`fentanyl solution overdose demonstrated
`that all dogs were moderately or severely sedated (Savides et al.,
`2012). In addition, the objective, continuous response variables
`of rectal body temperature and HR were collected 5 min before,
`10 min after, and 40 min after each naloxone dose. Additional
`sedation, rectal
`temperature, and HR measurements were
`collected at )1, 0, 14, 15, 24, 26, and 28 h following
`transdermal
`fentanyl solution administration. Venous blood
`samples for plasma fentanyl and naloxone concentrations were
`also collected from each dog at 0 (prior to transdermal fentanyl
`solution administration), 16 (prior to the 1st naloxone admin-
`istration), 20.083 (5 min following
`the 5th naloxone
`administration), and 24 (1 h following the last naloxone
`administration) hours post-transdermal fentanyl solution admin-
`istration directly into sodium heparin blood collection tubes.
`Plasma was harvested by centrifugation at 1500 g for 10 min at
`5 °C. Plasma samples were stored at )70 °C until analysis. All
`
`Ó 2012 Blackwell Publishing Ltd
`Nalox1030
`Nalox-1 Pharmaceuticals, LLC
`Page 2 of 7
`
`

`

`Naloxone reversal of transdermal fentanyl solution 47
`
`detection was conducted using positive ionization mode and
`the transitions 337.2 m ⁄ z fi 188.3 m ⁄ z
`monitoring of
`for
`fentanyl, 342.2 m ⁄ z fi 188.3 m ⁄ z for the IS fentanyl-d5, m ⁄ z
`328.2 fi m ⁄ z 310.2 for naloxone, and m ⁄ z 342.2 fi m ⁄ z 324.2
`for the IS naltrexone. Standard curves were determined using
`linear and quadratic regression for fentanyl and naloxone,
`respectively, with 1 ⁄ x2 weighting using Watson v7.0.0.01
`(Thermo Fisher Scientific), where x is the nominal sample
`concentration. Typical squared correlation coefficient (R2) values
`were 0.9972 and 0.9964 for fentanyl and naloxone, respec-
`tively. Concentration calculations were based on the peak area
`ratios of fentanyl to fentanyl-d5 and of naloxone to naltrexone
`for fentanyl and naloxone, respectively. The intra- and inter-
`assay precision (i.e., coefficient of variation) was £ 8.6%, and
`the accuracy (i.e., relative error) ranged from )4.2% to 6.0%
`for both analytes. The lower limit of quantification (LLOQ)
`was 0.1 and 1.0 ng ⁄ mL for plasma fentanyl and naloxone,
`respectively.
`
`Statistical methods
`
`The sedation assessments were analyzed using a generalized
`linear repeated measures mixed effects model. The logit trans-
`formation of the mean probability of sedation, li, for the ith
`subject was linearly related to time as follows:
`
`
`l
`1 l
`i
`

`logit l
`
`i
`

`
`8>>>>>><
`>>>>>>:
`
`Þ  log
`þ bi
`b
`B
`þ b
`b
`B
`þ b
`b
`B
`þ b
`b
`B
`þ b
`b
`B
`
`i
`
`þ bi
`þ b40
`þ b160
`þ bi
`
`N
`
`N
`
`þ bi
`þ bi
`
`F
`
`F
`
`F
`
`F
`
`if t  0
`if 0 < t  16
`if 16 < t  24 and Dose ¼ 40 lg=kg
`if 16 < t  24 and Dose ¼ 160 lg=kg
`if t > 24
`
`N
`
`3
`
`N
`
`where t is the nominal time of observation in hours, bB, bF, b40
`N ,
`N , are the baseline, fentanyl, 40 lg ⁄ kg i.m. naloxone,
`and b160
`and 160 lg ⁄ kg i.m. naloxone fixed effect terms, respectively.
`Additionally, bi is the subject-specific random effect term that is
`normally distributed with mean 0 and variance r2. The overall
`narcotic reversal effect of i.m. naloxone administered at hourly
`intervals on sedation was tested with the null hypothesis of
` b40
`þ 2
` b160
` 0 vs. the alternative hypothesis 1
` b40

` b160
`N
`3
`
`
`N < 0. The unequal weighting of b40N and b160N was used to
`account for the unbalanced design of the study (n = 8 and
`n = 16 for the 40 and 160 lg ⁄ kg naloxone doses, respectively).
`The narcotic reversal effectiveness of each naloxone dose was
`‡ 0, b160
`‡ 0 vs. the
`tested with the null hypotheses that b40
`N
`N
`
`N < 0, b160N < 0. To test
`alternative hypotheses that b40
`the
`additional effect of 160 lg ⁄ kg vs. a 40 lg ⁄ kg i.m. naloxone
`doses administered at hourly intervals, the null hypothesis of
`‡ b40
`b160
`N was tested against the alternative hypothesis that
`N
`
`N < b40N . As 100% of dogs in both group 1 and 2 were sedated
`b160
`during the time intervals 0 < t £ 16 and t > 24, a large
`correlation (near )1.00) existed between bF and b40
`
`N , b160N ,
`resulting in large standard errors of the parameters estimates.
`The large degree of correlation results in many combinations of
`
`13
`
`23
`
`Nalox1030
`Nalox-1 Pharmaceuticals, LLC
`Page 3 of 7
`
`procedures were approved by the local Institutional Animal Care
`and Use Committee.
`
`Plasma sample analysis
`
`Plasma fentanyl and naloxone concentrations were analyzed by
`liquid chromatography–tandem mass spectrometry. In brief, a

`stock solution of fentanyl (Cerilliant
`, Round Rock, TX, USA)

`) was diluted in
`and a stock solution of naloxone (Cerilliant

`, Morristown,
`50:50 methanol (Honeywell Burdick & Jackson
`NJ, USA):water (Milli-Q; Millipore Corp., Billerica, MA, USA) to a
`25 and 250 lg ⁄ mL working solution, respectively. Control dog
`plasma (Bioreclamation Inc., Hicksville, NY, USA) was then
`serially diluted with the working solution to create standard
`curve samples ranging from 0.1 to 10 ng ⁄ mL of fentanyl and 1
`to 1000 ng ⁄ mL naloxone. Additionally, an internal standard

`(IS) working solution of fentanyl-d5 (Cerilliant
`) and naltrexone

`) at concentrations 200 and 2000 ng ⁄ mL, respec-
`(Cerilliant
`tively, was prepared in 50:50 methanol ⁄ water. A 100 lL each
`of sample, standard, quality control, or control blank was
`aliquoted directly into a 96-well block, and 20 lL of the IS
`working solution was added to all wells except for the control
`blanks, which instead had 20 lL of 50:50 methanol ⁄ water were
`added and vortexed. Four hundred microliter of 5% acetic acid
`(Mallinckrodt Baker, Phillipsburg, NJ, USA) in water was then
`added to each well, and the samples were vortexed again
`followed by centrifugation at 4 °C. Solid-phase extraction (SPE)

`then proceeded using Bond Elut
`96 Certify, 50 mg sample
`extraction blocks (Varian Corp., Palo Alto, CA, USA), and a
`Tomtec Quadra-96 Model 320 (Tomtech, Hamden, CT, USA).
`Sample blocks were conditioned twice with 400 lL of methanol,
`followed by equilibration with two 400-lL volumes of Milli-Q
`water and equilibration with two 400-lL volumes of 5% acetic
`acid in water. Samples were then transferred to the SPE block
`and slowly aspirated. The SPE block was then washed twice with
`400 lL of 5% acetic acid in water followed by a two washes of
`400 lL of methanol. Samples were slowly eluted from the SPE
`block with two 300-lL volumes of 2% ammonium hydroxide
`(EMD Biosciences, Darmstadt, Germany) in acetonitrile (Honey-

`well Burdick & Jackson
`) and evaporated to dryness before
`reconstitution with 200 lL of 1% formic acid (EMD Biosciences)
`in acetonitrile.
`Reconstituted samples were quantified using an API 3000
`triple quadrupole mass spectrometer (Applied BioSystems ⁄ MDS
`SCIEX, Foster City, CA, USA) with peak area integration
`conducted using Analyst Software v 1.4 (Applied BioSys-
`tems ⁄ MDS SCIEX) data acquisition system. HPLC separation
`was achieved using a Thermo Betasil Silica-100 column
`(50 · 3 mm, 5 lm) (Thermo Fisher Scientific, Waltham, MA,
`USA) with the flow rate set at 0.5 mL ⁄ min. Mobile phase A
`consisted of 1% formic acid in water and mobile phase B
`consisted of 1% formic acid in acetonitrile. The mobile phase
`gradient started at 90% mobile phase B from 0.0 to 1.0 min,
`switched from 90% to 70% mobile phase B from 1.0 to 1.5 min,
`and switched back from 70% to 90% mobile phase B at 2.5 min.
`The injection volume was 10 lL, and mass spectrometer
`
`Ó 2012 Blackwell Publishing Ltd
`
`

`

`48 K. J. Freise et al.
`
`parameter estimates giving almost identical fits to the data. To
`alleviate this issue, the bF parameter was fixed to the initially
`estimated value (i.e., the estimate when all five terms in the
`model were simultaneously estimated), and then, the statistical
`analysis was conducted to determine the effect of naloxone on
`sedation, conditional on the fixed value of bF. The bF parameter
`was chosen to be fixed as it is already known that fentanyl has
`sedative effects (Freise et al., 2012b; Adams, 2001; Plumb,
`2002) and because the interest of the study was the effect of
`naloxone, not the effect of fentanyl. A sensitivity analysis was
`subsequently conducted to determine the effect on the statistical
`conclusions of changing the value of bF to other reasonable
`values.
`The body temperature and HR were analyzed with a linear
`repeated measures mixed effects model with dose, nominal time,
`and dose by nominal time interaction terms as fixed effects and
`subject as a random effect in the model. The covariance structure
`in the repeated measures analysis was investigated using three
`structural assumptions, namely compound symmetry, first-order
`autoregressive, and heterogeneous first-order autoregressive.
`The assumption that gave the minimum value of the Akaike’s
`Information Criterion was selected for the final model (Akaike,
`1974). For both body temperature and HR, the first-order
`autoregressive model was selected. The overall narcotic reversal
`effect of
`i.m. naloxone administered at hourly intervals was
`tested with a null hypothesis of lN £ lF, vs. the alternative
`hypothesis lN > lF, where lN is the mean body temperature or
`HR during the naloxone treatment time period (16 < t £ 24)
`and lF is the mean body temperature or HR during the fentanyl
`only time period (t = 14, 15, 15.917, 26, 28). To test the
`additional effect of 160 lg ⁄ kg vs. a 40 lg ⁄ kg i.m. naloxone
`doses administered at hourly intervals, the null hypothesis of
`£ l40
`l160
`N was tested against the alternative hypothesis of
`N
`l160
`
`
`N > l40N , where l40N and l160
`are the mean body temperatures
`N
`or HRs during the naloxone treatment time period for the 40 and
`160 lg ⁄ kg i.m. naloxone dose groups, respectively.
`All statistical analyses and calculations were conducted in
`SAS (version 9.1.3 Service Pack 4; SAS Institute Inc., Cary, NC,
`USA). The sedation scores were analyzed using the NLMIXED
`procedure, and the body temperatures and HR were analyzed
`using the MIXED procedure. Statistically significant differences
`the a = 0.05 probability of a type I
`were determined at
`experiment-wise error. To control the experiment-wise error
`rate, the unadjusted P-values were corrected using the step-
`
`down Bonferonni method for multiple tests on each response
`variable (Holm, 1979). Specific hypotheses were tested using the
`ESTIMATE statement in SAS and unadjusted P-values con-
`structed using a Student’s t-test.
`
`RESULTS
`
`The plasma naloxone and fentanyl concentrations are displayed
`in Table 1. Plasma fentanyl concentrations were below the LLOQ
`prior to dosing in all dogs, and the mean fentanyl concentrations
`ranged from 4.60 to 6.53 ng ⁄ mL across both groups from 16
`through 24 h following the administration of a fivefold overdose
`(13 mg ⁄ kg) of
`transdermal
`fentanyl
`solution. The plasma
`naloxone concentrations were also below the LLOQ prior to
`i.m. naloxone dose administration in all dogs. At 5 min following
`the 5th naloxone dose administration (20.083 h), the plasma
`naloxone concentrations were 10.4 ± 0.238 (mean ± standard
`error) and 34.7 ± 1.76 ng ⁄ mL in the 40 and 160 lg ⁄ kg i.m.
`naloxone dose groups, respectively. At 24 h, the mean plasma
`naloxone concentrations had dropped substantially from the
`previous peaks in both groups, consistent with its known short
`duration of action and rapid clearance (Veng-Pedersen et al.,
`1995; Adams, 2001; Plumb, 2002). No seizures or other adverse
`affects of naloxone administration were observed in any dogs.
`The observed proportions of dogs sedated vs.
`time are
`displayed in Fig. 1 for both the 40 and 160 lg ⁄ kg naloxone
`dose groups. As can be observed, the baseline, pretransdermal
`fentanyl solution administration proportion of sedated dogs is
`near 0.0. For nonapparent reasons, six dogs were scored as
`sedated at the time of transdermal fentanyl solution application
`resulting in the proportion of sedated dogs being 0.4 and 0.2 in
`the 40 and 160 lg ⁄ kg groups, respectively, at time 0. Following
`a fivefold overdose of transdermal
`fentanyl solution, all dogs
`were sedated prior to naloxone administration (i.e., at 14, 15,
`15.917 h). The administration of either 40 or 160 lg ⁄ kg i.m.
`naloxone at hourly intervals reduced the proportion of sedated
`dogs. The mean proportion of sedated dogs from 16 through
`24 h for the 40 and 160 lg ⁄ kg dose groups was 0.698 and
`0.438, respectively. Additionally, all dogs were determined to be
`sedated at least once from 16 through 24 h in both groups. The
`mean proportion of sedated dogs returned to 1.0 following
`cessation of the hourly i.m. naloxone administrations for both
`groups by 26 h.
`
`Table 1. Plasma fentanyl and naloxone concentrations by treatment group
`
`Plasma fentanyl conc. (ng ⁄ mL)
`40 lg ⁄ kg i.m. naloxone
`160 lg ⁄ kg i.m. naloxone
`
`Plasma naloxone conc. (ng ⁄ mL)
`40 lg ⁄ kg i.m. naloxone
`160 lg ⁄ kg i.m. naloxone
`
`Time (h)
`
`Mean
`
`Standard error
`
`Mean
`
`Standard error
`
`Mean
`
`Standard error
`
`Mean
`
`Standard error
`
`0
`16
`20.083
`24
`
`<LLOQ
`4.60
`5.92
`5.42
`
`–
`0.537
`0.873
`0.919
`
`<LLOQ
`5.46
`5.80
`6.53
`
`–
`0.295
`0.358
`0.568
`
`<LLOQ
`<LLOQ
`10.4
`2.78
`
`–
`–
`0.238
`0.201
`
`<LLOQ
`<LLOQ
`34.7
`12.8
`
`–
`–
`1.76
`0.848
`
`<LLOQ, Less than lower limit of quantification for all subjects.
`
`Ó 2012 Blackwell Publishing Ltd
`Nalox1030
`Nalox-1 Pharmaceuticals, LLC
`Page 4 of 7
`
`

`

`Naloxone reversal of transdermal fentanyl solution 49
`
`40 (cid:1106)g/kg
`
`160 (cid:1106)g/kg
`
`38.5
`
`37.5
`
`36.5
`
`35.5
`
`34.5
`
`Temperature (°C)
`
`16
`
`20
`
`24
`
`28
`
`–4
`
`0
`
`4
`
`8
`
`16
`
`20
`
`24
`
`28
`
`12
`Time (h)
`
`40 (cid:1106)g/kg
`
`160 (cid:1106)g/kg
`
`1
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`Proportion sedated
`
`0
`
`–4
`
`0
`
`4
`
`8
`
`12
`Time (h)
`
`Fig. 1. Observed proportion of dogs sedated vs. time by i.m. naloxone
`treatment group.
`
`Fig. 2. Mean rectal body temperature vs. time by i.m. naloxone
`treatment group. Bars represent the standard error.
`
`40 (cid:1106)g/kg
`
`160 (cid:1106)g/kg
`
`140
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`Heart rate (bpm)
`
`–4
`
`0
`
`4
`
`8
`
`12
`Time (h)
`
`16
`
`20
`
`24
`
`28
`
`Fig. 3. Mean heart rate vs. time by i.m. naloxone treatment group. Bars
`represent the standard error.
`
`101 ± 3.31 bpm prior to transdermal fentanyl solution admin-
`istration to 64.2 ± 3.04 bpm following transdermal
`fentanyl
`solution administration (i.e., at time 14, 15, and 15.917 h).
`During i.m. naloxone reversal (i.e., from 16 through 24 h), the
`HR across both groups returned to the prefentanyl administra-
`tion HR with a value of 101 ± 2.41 bpm and then dropped
`again to an overall mean of 83.1 ± 3.31 bpm following
`termination of naloxone administration. The mean HR during
`time period (lN) was 28.9 ± 1.78 bpm
`naloxone treatment
`higher than the mean during the fentanyl only time period (lF)
`(P < 0.001). Finally, during the naloxone treatment time period,
`the HR was 9.97 ± 5.11 bpm higher in the 160 lg ⁄ kg i.m.
`naloxone dose group (104 ± 2.95 bpm) than in the 40 lg ⁄ kg
`i.m. naloxone dose group (94.4 ± 4.17 bpm, P = 0.0258),
`further
`indicating greater narcotic reversal effect of
`the
`160 lg ⁄ kg i.m. naloxone dosage.
`
`DISCUSSION
`
`This study demonstrates that the opioid-induced effects of up to a
`fivefold overdose of transdermal fentanyl solution in dogs can be
`successfully reversed through administrations of either 40 or
`160 lg ⁄ kg i.m. naloxone. Naloxone is an approved opioid
`antagonist for use in the dog where the recommended initial dose
`
`Nalox1030
`Nalox-1 Pharmaceuticals, LLC
`Page 5 of 7
`
`The overall effect of naloxone on reversal of the sedative effects
`of transdermal
`fentanyl solution was statistically significant
`(P < 0.001), as was the individual effect of
`the 40 and
`160 lg ⁄ kg i.m. naloxone reversal regimens (P < 0.001 for both
`regimens). The analysis also indicated that there was significant
`subject-to-subject variability in the sedation response (i.e., the
`probability that r2 > 0 was < 0.05). Furthermore, the reversal
`affect of the 160 lg ⁄ kg i.m. naloxone dose was significantly
`the 40 lg ⁄ kg i.m. naloxone dose
`greater
`than that
`for
`(P = 0.0132). The odds of a subject being sedated with a
`160 lg ⁄ kg i.m. naloxone dose was 0.353 (95% confidence
`interval [0.0327–0.674]) fold that of a 40 lg ⁄ kg i.m. naloxone
`dose. Owing to the high degree of correlation (near )1.00)
`
`
`between bF and b40N , b160N , the value of bF was fixed to the initial
`estimate of 11.5. Varying the fixed value of bF from 1 to 30 had
`no affect on the hypothesis test results (accurate numerical
`integration of the likelihood could not be achieved for values of
`bF > 30),
`indicating that the results are robust to different
`reasonable fixed values of bF.
`The mean rectal body temperatures vs. time by i.m. naloxone
`dose group are displayed in Fig. 2. The rectal body tempera-
`tures across both groups dropped from 38.4 ± 0.0976 °C prior
`to transdermal
`fentanyl solution administration to 35.1 ±
`0.0884 °C following transdermal
`fentanyl solution treatment
`(i.e., at time 14, 15, and 15.917 h). During i.m. naloxone
`reversal (i.e.,
`from 16 through 24 h), the body temperature
`across both groups was 37.7 ± 0.0578 °C. By 26 and 28 h
`the body temperatures returned to near prenaloxone adminis-
`tration values with an overall mean of 35.9 ± 0.0976 °C. The
`mean body temperature during naloxone treatment
`time
`period (lN) was 2.19 ± 0.0638 °C higher than the mean
`during the fentanyl only time period (lF) (P < 0.001). Addi-
`tionally, during the naloxone treatment time period, the body
`temperature was 0.412 ± 0.123 °C higher in the 160 lg ⁄ kg
`i.m. naloxone dose group (37.8 ± 0.0708 °C)
`than in the
`40 lg ⁄ kg
`(37.4 ± 0.100 °C,
`i.m. naloxone
`dose
`group
`P < 0.001),
`indicating greater narcotic reversal effect of the
`higher i.m. naloxone dose.
`The mean HRs vs. time by i.m. naloxone dose group are
`displayed in Fig. 3. The HRs across both groups dropped from
`
`Ó 2012 Blackwell Publishing Ltd
`
`

`

`50 K. J. Freise et al.
`
`is 40 lg ⁄ kg administered by i.m., i.v., or subcutaneous injection
`followed by repeated doses, as needed, with at least 2–3 min
`between doses. It has been previously demonstrated to have rapid
`onset of action within 1–2 min of injection and a 45-min–3-h
`duration of action following a 30 lgÆkg ⁄ min fentanyl constant
`rate infusion (CRI) (Veng-Pedersen et al., 1995; Plumb, 2002).
`Likewise in the current study, renarcotization of dogs to at or near
`prereversal
`levels following a fivefold overdose of
`fentanyl
`occurred between 1 and 3 h after termination of the hourly
`naloxone dosage regimens. Naloxone is generally considered to be
`a safe agent with a wide therapeutic window; however, extremely
`high doses may cause seizures through c-aminobutyric acid
`receptor antagonism (Adams, 2001; Plumb, 2002). In the present
`study, i.m. naloxone administration was safe and the reversal of
`the sedative, hypothermic, and bradycardic effects of transdermal
`fentanyl solution occurred within minutes. Although both
`naloxone regimens were effective, the hourly 160 lg ⁄ kg i.m.
`naloxone reversal regimen was more effective at reversing the
`opioid-induced effects of an overdose.
`Sedation was recorded in six of 24 dogs in both treatment
`groups immediately prior to transdermal
`fentanyl solution
`administration, even though plasma fentanyl concentrations
`were not detectable. Therefore, the pretransdermal
`fentanyl
`solution administration sedation observations were most likely
`due to the subjective nature of sedation assessments. For
`example, the sedation scale that was utilized did not distinguish
`between mild, moderate, and severe sedation and a possibility
`was that blinded assessors could have scored relaxed dogs as
`sedated. Because of the subjective nature of sedation assessments
`and the numerical difficulties that required one of the model
`parameters to be fixed,
`the objective continuous response
`variables of body temperature and HR were also analyzed. The
`analysis of body temperature and HR response data confirm the
`conclusions from the sedation assessments, that both the 40 and
`160 lg ⁄ kg i.m. naloxone regimens were effective at reversing
`the opioid-induced effects of transdermal fentanyl solution and
`that the 160 lg ⁄ kg reversal regimen was more effective.
`Intramuscular naloxone rapidly reversed the effects of fentanyl
`overdose within minutes as clearly documented from the sedation,
`body temperature, and HR observations 10 min following the first
`naloxone administration. These results support the idea that rapid
`results can be achieved by i.m. injection without the need for direct
`i.v. access. The duration of a single naloxone injection is also
`illustrated by the peak and trough nature of the reversal between
`hourly injections. This phenomenon is likely reflective of the rapid
`absorption and elimination of naloxone which has a terminal
`elimination half-life of 1.19 h in the dog (Pace et al., 1979). The
`short duration of naloxone to reverse a fivefold overdose is
`illustrated in the 40 lg ⁄ kg group, whereby frequently all dogs
`were sedated immediately prior to the next naloxone administra-
`tion. Body temperatures nearly returned to the pretransdermal
`fentanyl solution administration temperatures, particularly in the
`160 lg ⁄ kg treatment group. Across both groups, the mean body
`temperature during the naloxone administration time period was
`only 0.7 °C below the pretransdermal fentanyl solution admin-
`istration mean body temperature illustrating near complete
`
`reversal. Likewise, naloxone administration returned the mean
`HR to approximately the pretransdermal fentanyl solution mean
`HR, indicating near complete reversal of the bradycardia.
`Fentanyl-induced respiratory depression and its reversal by
`naloxone were not assessed in this study as a previous dog safety
`study using up to a fivefold overdose of transdermal fentanyl
`solution demonstrated that respiratory rates were maximally
`decreased only 30% (Savides et al., 2012). Furthermore, other
`studies have demonstrated that plasma fentanyl concentrations
`as high as approximately 80 ng ⁄ mL reduce the respiratory rate
`by only approximately 11 breaths ⁄ min (50%) in spontaneously
`breathing dogs (Arndt et al., 1984). Respiratory rate, oxygen
`consumption, and blood gases (pCO2, pO2, and pH) do not change
`further as concentrations increase above 100 ng ⁄ mL. Another
`study of fentanyl in dogs using a transdermal patch confirmed by
`blood gas analysis that sustained steady-state plasma fentanyl
`concentrations of approximately 2 ng ⁄ mL do not cause hypo-
`ventilation (Welch et al., 2002). When taken together, the data
`indicate that respiratory depression is a safety aspect of limited
`concern following fentanyl administration to dogs.
`The shorter duration of action of naloxone relative to
`transdermal fentanyl solution may necessitate repeat injections
`until an overdose is
`satisfactorily treated. Application of
`transdermal fentanyl solution to the skin results in sequestration
`of fentanyl into the stratum corneum and sustained absorption
`characterized by flip-flop kinetics with a half-life of approxi-
`mately 70 h (Freise et al., 2012a,b). As a result of the prolonged
`absorption into systemic circulation over a period of days, in

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket